Recombinant Rat UDP-glucuronosyltransferase 2B17 (Ugt2b17)

Shipped with Ice Packs
In Stock

Product Specs

Form
Lyophilized powder
Note: While we prioritize shipping the format currently in stock, please specify your format preference in order notes for customized preparation.
Lead Time
Delivery times vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Note: Standard shipping includes blue ice packs. Dry ice shipping requires prior arrangement and incurs additional charges.
Notes
Avoid repeated freeze-thaw cycles. Store working aliquots at 4°C for up to one week.
Reconstitution
Centrifuge the vial briefly before opening to consolidate the contents. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. For long-term storage, we recommend adding 5-50% glycerol (final concentration) and aliquoting at -20°C/-80°C. Our standard glycerol concentration is 50% and can be used as a reference.
Shelf Life
Shelf life depends on storage conditions, buffer composition, temperature, and protein stability. Generally, liquid formulations have a 6-month shelf life at -20°C/-80°C, while lyophilized formulations have a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquot for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag type is determined during manufacturing.
The tag type is determined during production. If you require a specific tag, please inform us, and we will prioritize its development.
Synonyms
Ugt2b17; Ugt2b3; Ugt2b5; UDP-glucuronosyltransferase 2B17; UDPGT 2B17; UGT2B17; 17-beta-hydroxysteroid-specific UDPGT; RLUG38; Testosterone, dihydrotestosterone, and beta-estradiol-specific UDPGT; UDP-glucuronosyltransferase 2B5; UDPGT 2B5; UDPGTr-3
Buffer Before Lyophilization
Tris/PBS-based buffer, 6% Trehalose.
Datasheet
Please contact us to get it.
Expression Region
24-530
Protein Length
Full Length of Mature Protein
Species
Rattus norvegicus (Rat)
Target Names
Ugt2b17
Target Protein Sequence
GKVLVWPMEFSHWMNIKTILDELVQRGHEVTVLKPSAYYVLDPKKSPDLKFETFPTSVSK DELENYFIKLVDVWTYELQRDTCLSYSPLLQNMIDGFSDYYLSLCKDTVSNKQLMAKLQE SKFDVLLSDPVAACGELIAEVLHIPFLYSLRFSPGYKIEKSSGRFILPPSYVPVILSGMG GPMTFIDRVKNMICTLYFDFWFHMFNAKKWDPFYSEILGRPTTLAETMGKAEMWLIRSYW DLEFPHPTLPNVDYIGGLQCRPPKPLPKDMEDFVQSSGEHGVVVFSLGSMVSSMTEEKAN AIAWALAQIPQKVLWKFDGKTPATLGPNTRVYKWLPQNDLLGHPKTKAFVTHSGANGVYE AIYHGIPMVGIPMFGEQHDNIAHMVAKGAAVTLNIRTMSKSDLFNALKEIINNPFYKKNA VWLSTIHHDQPMKPLDKAVFWIEFVMRHKGAKHLRPLGHDLPWYQYHSLDVIGFLLTCSA VIAVLTVKCFLFIYRLFVKKEKKMKNE
Uniprot No.

Target Background

Function

Recombinant Rat UDP-glucuronosyltransferase 2B17 (UGT2B17) is a phase II biotransformation enzyme that catalyzes the conjugation of lipophilic substrates with glucuronic acid. This process enhances the water solubility of metabolites, facilitating their excretion via urine or bile. UGT2B17 specifically catalyzes the glucuronidation of endogenous steroid hormones, including androgens (e.g., epitestosterone, androsterone) and estrogens (e.g., estradiol, epiestradiol).

Database Links
Protein Families
UDP-glycosyltransferase family
Subcellular Location
Endoplasmic reticulum membrane; Single-pass membrane protein.

Q&A

How does recombinant rat UGT2B17 compare to other rat UGT isoforms in glucuronidation assays?

Recombinant rat UGT2B17 exhibits distinctive substrate preferences compared to other rat UGT isoforms. When screening various rat UGT isoforms, researchers typically observe different glucuronidation patterns and kinetic parameters for diverse substrates. Similar to how human UGT2B7 has been shown to exhibit higher activity than other UGTs for certain substrates like R(+)-NAF and S(-)-NAF , rat UGT isoforms demonstrate varying activities. For proper comparative analysis, researchers should conduct enzyme kinetic studies with multiple substrates across various UGT isoforms under standardized conditions, measuring parameters such as Km, Vmax, and intrinsic clearance to characterize the relative contributions of each isoform.

What experimental conditions are optimal for assessing recombinant rat UGT2B17 activity in vitro?

Optimal experimental conditions for assessing recombinant rat UGT2B17 activity typically include:

  • Incubation medium: Tris-HCl buffer (usually 50-100 mM, pH 7.4-7.5)

  • Cofactor concentration: 4 mM UDP-glucuronic acid (UDPGA)

  • Enzyme activation: Treatment with alamethicin (5% of microsomal protein concentration) on ice for 20 minutes to ensure maximum activity

  • Substrate concentration: Ranging from 0.2-20 μM for kinetic assays, depending on substrate properties

  • Incubation time and protein concentration: Must be determined in preliminary experiments to ensure linearity of glucuronide formation

  • Temperature: 37°C for incubation

  • Reaction termination: Addition of cold methanol followed by centrifugation at 20,000 g for 20 min at 4°C

The final reaction mixture should contain less than 1% organic solvent to avoid enzyme inhibition. Analysis of glucuronides typically involves HPLC-UV or LC-MS/MS methodologies.

What are the recommended procedures for expressing and purifying recombinant rat UGT2B17?

Recombinant rat UGT2B17 expression and purification typically follows these protocols:

  • Expression system selection: Baculovirus-infected insect cells (Sf9) or mammalian cell lines (HEK293, CHO) are preferred as they provide proper post-translational modifications.

  • Vector design: The full-length rat UGT2B17 cDNA should be cloned into an appropriate expression vector with a 6×His-tag or similar affinity tag for purification.

  • Expression optimization:

    • Transfection/infection conditions optimization

    • Expression verification via Western blot

    • Small-scale expression tests to determine optimal harvest time

  • Purification protocol:

    • Cell lysis using mild detergents (0.5-1% Triton X-100)

    • Membrane fraction isolation by ultracentrifugation

    • Solubilization of membrane proteins

    • Affinity chromatography using nickel-NTA resin

    • Size exclusion chromatography for higher purity

  • Enzyme activity verification: Testing with known substrates to confirm functionality after purification

For reliable results, it's crucial to verify both the purity via SDS-PAGE and the functional integrity through activity assays with model substrates.

How should researchers design kinetic assays for recombinant rat UGT2B17 to ensure reproducible results?

Designing robust kinetic assays for recombinant rat UGT2B17 requires careful attention to several methodological aspects:

  • Preliminary assays:

    • Determine linear range for both reaction time and protein concentration

    • Enzyme stability assessment under assay conditions

  • Substrate concentration range:

    • Select 7-9 substrate concentrations spanning from 0.1×Km to 10×Km

    • For substrates with unknown Km, use logarithmic concentration spacing (e.g., 0.2, 0.5, 1, 2, 5, 10, 20 μM)

  • Incubation conditions standardization:

    • Buffer composition and pH consistency

    • Fixed temperature (37°C)

    • Controlled UDPGA concentration (typically 4 mM)

  • Data analysis approach:

    • Fit data to appropriate kinetic models:

      • Michaelis-Menten equation: v = Vmax × [S]/(Km + [S])

      • Substrate inhibition model: v = Vmax × [S]/[Km + [S] × (1 + [S]/Ki)]

    • Determine kinetic parameters (Km, Vmax, CLint)

  • Quality control measures:

    • Include positive controls (known substrates)

    • Run all assays in triplicate

    • Include inter-day and intra-day validation

Table 1: Example of Kinetic Parameter Determination Setup for Recombinant Rat UGT2B17

ParameterSpecification
Protein concentration0.01-0.1 mg/mL (optimized for linearity)
Incubation time20-30 minutes (within linear range)
Substrate concentrations0.2, 0.5, 1, 2, 5, 10, 20 μM
Buffer50-100 mM Tris-HCl, pH 7.4-7.5
Cofactor4 mM UDPGA
Temperature37°C
ReplicatesMinimum triplicate experiments
Analysis methodHPLC-UV or LC-MS/MS

What analytical methods are most suitable for detecting and quantifying rat UGT2B17-mediated glucuronidation products?

The most suitable analytical methods for detecting and quantifying glucuronidation products include:

  • HPLC-UV/Vis detection:

    • Suitable for substrates with chromophores

    • Requires relatively higher concentrations of metabolites

    • Advantages: Accessible equipment, straightforward method development

    • Limitations: Lower sensitivity, potential co-elution issues

  • LC-MS/MS analysis:

    • Gold standard for glucuronide detection and quantification

    • Offers structural confirmation through mass spectrometry

    • Detection of deprotonated glucuronide metabolites (typically M+176 m/z compared to parent compound)

    • Multiple reaction monitoring (MRM) for enhanced sensitivity and specificity

  • Radiometric assays:

    • Using 14C-UDPGA as cofactor

    • Highly sensitive but requires special handling

  • Method validation requirements:

    • Linearity (r² > 0.99)

    • Precision (CV < 15%)

    • Accuracy (within 85-115% of nominal concentration)

    • Lower limit of quantification determination

    • Matrix effect evaluation

For comprehensive metabolite profiling, a combination of HPLC separation with both UV detection and mass spectrometric analysis provides the most robust approach for identification and quantification of glucuronide conjugates.

How do genetic polymorphisms in rat UGT2B17 influence enzyme activity compared to the well-documented human UGT2B17 variations?

While human UGT2B17 is characterized by a common gene deletion polymorphism with significant functional consequences , less is documented about genetic variations in rat UGT2B17. The human UGT2B17 gene deletion is associated with:

  • Altered drug pharmacokinetics:

    • 25-fold higher systemic exposure to some drugs in subjects with two gene copies compared to homozygous deletion carriers

    • Significant impact on the metabolism of drugs like exemestane and vorinostat

  • Functional consequences:

    • Changes in steroid hormone metabolism, particularly testosterone glucuronidation

    • Potentially altered risk for steroid-dependent cancers

For rat UGT2B17 research, investigators should:

  • Sequence the UGT2B17 gene in different rat strains to identify potential polymorphisms

  • Generate recombinant variants of the identified polymorphisms

  • Compare enzyme kinetics (Km, Vmax, CLint) of variants using model substrates

  • Assess expression levels in different tissues across rat strains

  • Conduct comparative studies between species-specific variants to understand evolutionary conservation of important functional domains

This comparative approach would provide insights into species differences in UGT2B17 regulation and function that are crucial for translational research.

What approaches can be used to investigate substrate binding mechanisms and catalytic activity of recombinant rat UGT2B17?

Advanced investigation of substrate binding and catalytic mechanisms requires multidisciplinary approaches:

  • Protein structure analysis:

    • Homology modeling based on available UGT crystal structures

    • Molecular docking simulations with known substrates

    • Site-directed mutagenesis of predicted binding residues

  • Enzyme kinetics with multiple substrates:

    • Determination of substrate inhibition patterns (competitive, non-competitive, uncompetitive)

    • Analysis using substrate inhibition models: v = Vmax × [S]/[Km + [S] × (1 + [S]/Ki)]

    • Evaluation of stereoselectivity (as seen with human UGT2B17 for 17β-estradiol)

  • Spectroscopic techniques:

    • Fluorescence spectroscopy to monitor conformational changes upon substrate binding

    • Circular dichroism to assess secondary structure alterations

    • Isothermal titration calorimetry for binding thermodynamics

  • Chemical modification approaches:

    • Selective chemical modification of key amino acid residues

    • Identification of essential catalytic residues

    • pH-rate profiles to determine ionizable groups involved in catalysis

Table 2: Methods for Investigating Rat UGT2B17 Substrate Binding and Catalysis

ApproachTechniqueInformation Obtained
ComputationalHomology modelingPredicted 3D structure
Molecular dockingSubstrate binding orientation
MD simulationsDynamics of enzyme-substrate interactions
BiochemicalEnzyme kineticsKm, Vmax, CLint, substrate inhibition patterns
Inhibition studiesCompetitive vs. non-competitive mechanisms
pH-dependenceIonizable groups in catalytic site
BiophysicalFluorescence spectroscopyConformational changes upon binding
Circular dichroismSecondary structure changes
Isothermal titration calorimetryBinding thermodynamics
Molecular BiologySite-directed mutagenesisCritical binding/catalytic residues
Chimeric enzymesDomain-specific functions

How can researchers effectively compare glucuronidation profiles between recombinant rat UGT2B17 and human UGT2B17 for translational research?

Effective comparison of glucuronidation profiles between species requires systematic experimental design:

  • Standardized expression systems:

    • Expression of both enzymes in identical systems (e.g., baculovirus-infected insect cells)

    • Quantification of expression levels for normalization

    • Verification of proper folding and post-translational modifications

  • Comprehensive substrate panel screening:

    • Testing both enzymes against a diverse panel of substrates (steroids, drugs, xenobiotics)

    • Determination of substrate specificity profiles

    • Identification of species-specific and shared substrates

  • Detailed enzyme kinetics:

    • Determination of kinetic parameters for selected substrates

    • Comparison of catalytic efficiency (Vmax/Km)

    • Analysis of stereoselectivity and regioselectivity patterns

  • Inhibition profiles:

    • Sensitivity to known UGT inhibitors

    • Species differences in inhibition constants (Ki values)

  • Physiologically relevant models:

    • Comparison using liver microsomes from both species

    • Scaling of in vitro data to predict in vivo clearance

    • Evaluation of the relative contribution to total glucuronidation in each species

This systematic approach facilitates better translation of preclinical rat data to human clinical scenarios, particularly important given that human UGT2B17 genetic polymorphisms can lead to unexpected pharmacokinetic outcomes and drug development failures .

What statistical approaches are appropriate for analyzing enzyme kinetic data from recombinant rat UGT2B17 studies?

Appropriate statistical approaches for UGT2B17 kinetic data analysis include:

  • Model selection for enzyme kinetics:

    • Eadie-Hofstee plots to identify the appropriate kinetic model

    • Akaike Information Criterion (AIC) for objective model selection

    • R-squared values for goodness of fit assessment

    • Selection between:

      • Simple Michaelis-Menten model: v = Vmax × [S]/(Km + [S])

      • Substrate activation model

      • Substrate inhibition model: v = Vmax × [S]/[Km + [S] × (1 + [S]/Ki)]

  • Parameter estimation:

    • Non-linear least squares regression using software like GraphPad Prism

    • Bootstrapping for confidence interval determination

    • Evaluation of parameter uncertainty

  • Comparative statistical analysis:

    • For comparing kinetic parameters between different conditions:

      • Student's t-test for comparing two conditions

      • ANOVA followed by post-hoc tests for multiple conditions

      • Kruskal-Wallis or Mann-Whitney tests for non-parametric data

      • Jonckheere-Terpstra trend tests for ordered effects

  • Correlation analysis:

    • Spearman or Pearson correlation for assessing relationships between parameters

    • Multiple linear regression for modeling complex relationships

When reporting kinetic parameters, always include both the point estimates (mean values) and measures of uncertainty (standard deviation, standard error, or confidence intervals), along with the number of replicates (typically triplicate experiments) .

How can researchers address data discrepancies when comparing recombinant rat UGT2B17 activity with native enzyme activity in rat liver microsomes?

Addressing discrepancies between recombinant and microsomal UGT2B17 activity requires systematic investigation:

  • Sources of potential discrepancies:

    • Expression system effects on post-translational modifications

    • Absence of membrane environment in some recombinant preparations

    • Different UGT isoforms contributing to microsomal activity

    • Protein-protein interactions present in microsomes but absent in recombinant systems

  • Quantitative approaches to resolve discrepancies:

    • Relative activity factor (RAF) determination

    • Quantification of UGT2B17 protein in microsomes using immunoquantification

    • Selective inhibition studies to isolate UGT2B17 contribution in microsomes

    • Correlation analysis between recombinant activity and microsomal activity across multiple substrates

  • Experimental strategies:

    • Use of chemical inhibitors like fluconazole to inhibit specific UGT isoforms

    • Immunoinhibition with UGT2B17-specific antibodies

    • Comparison across species and tissues to identify patterns (liver, kidney, intestinal microsomes)

    • Testing with known selective substrates for UGT2B17

  • Data normalization approaches:

    • Normalization to marker substrate activity

    • Protein expression level correction

    • Inter-system extrapolation factors

What are the key considerations when interpreting species differences between rat and human UGT2B17 for drug metabolism studies?

Key considerations for interpreting species differences include:

  • Structural and functional differences:

    • Sequence homology analysis between rat and human UGT2B17

    • Differences in substrate binding sites and catalytic residues

    • Species-specific post-translational modifications

  • Expression pattern differences:

    • Tissue distribution variations between species

    • Absolute expression level differences

    • Influence of sex, age, and disease state on expression

  • Genetic polymorphism considerations:

    • High prevalence of gene deletion in human UGT2B17

    • Effect on drug disposition (e.g., exemestane metabolism)

    • Potential rat strain differences in UGT2B17 genetics

  • Translational implications:

    • Quantitative scaling factors for predicting human pharmacokinetics

    • Identification of drugs with potential species-dependent metabolism

    • Development of correction factors for preclinical-to-clinical translation

  • Data interpretation framework:

    • Integration of in vitro, in silico, and in vivo data

    • Physiologically-based pharmacokinetic (PBPK) modeling

    • Consideration of compensatory mechanisms (redundancy with other UGTs)

Understanding these differences is critical since UGT2B17 variability has led to unexpected pharmacokinetic outcomes resulting in drug development failures, as documented with MK-7246 and PT2385 .

How do tissue-specific differences in UGT2B17 expression affect glucuronidation capacity in rats compared to humans?

Tissue-specific differences in UGT2B17 expression create distinct glucuronidation patterns:

  • Tissue distribution patterns:

    • Human UGT2B17 is highly expressed in liver, with significant expression also found in steroid-responsive tissues

    • Rat UGT2B17 tissue distribution may differ, requiring systematic quantification across tissues

  • Relative contribution to metabolism:

    • In humans, liver, kidney, and intestinal microsomes show different glucuronidation capacities for various substrates

    • Similar tissue-specific differences have been observed in rats

    • Comparisons of rat liver microsomes (RLM) and rat intestinal microsomes (RIM) show distinct kinetic parameters for substrates

  • Experimental approach for tissue comparison:

    • Preparation of microsomes from multiple tissues (liver, kidney, intestine)

    • Activity assays with the same substrate panel across tissues

    • Protein expression quantification via Western blot or LC-MS/MS

    • mRNA expression analysis via RT-qPCR

  • Physiological implications:

    • First-pass metabolism differences

    • Tissue-specific drug accumulation

    • Target tissue exposure to active compounds

Studies have shown that human UGT2B7 (another UGT isoform) is quantified as the most abundant UGT in liver and kidney with decreased levels in intestine , and similar tissue distribution analyses for UGT2B17 would provide valuable insights into metabolism patterns.

What methodologies are most effective for studying the impact of UGT2B17 genetic variations on drug metabolism across species?

Effective methodologies for studying UGT2B17 genetic variations include:

  • Genetic analysis approaches:

    • Whole genome sequencing of rat strains to identify natural variants

    • Targeted sequencing of UGT2B17 gene and regulatory regions

    • Copy number variation analysis (especially relevant given the common deletion in humans)

    • Haplotype determination

  • Functional genomics:

    • CRISPR/Cas9 gene editing to create rat models with specific UGT2B17 variants

    • Generation of humanized rat models expressing human UGT2B17 variants

    • Recombinant expression of variant alleles for in vitro characterization

  • Phenotypic characterization:

    • In vitro metabolism studies with liver microsomes from different rat strains

    • In vivo pharmacokinetic studies in rat strains with different UGT2B17 genotypes

    • Correlation of genotype with glucuronidation activity using marker substrates

  • Translational approaches:

    • Comparative analysis between rat models and human clinical data

    • PBPK modeling incorporating genetic variation data

    • Utilization of testosterone glucuronidation as a potential urinary biomarker

Table 3: Key Methodologies for Cross-Species UGT2B17 Variation Studies

MethodologyApplicationOutcome Measures
Genetic AnalysisSequencing of rat UGT2B17 geneIdentification of variants
Copy number variation analysisDetermination of gene dosage
Promoter region analysisRegulatory variants identification
Functional GenomicsCRISPR/Cas9 gene editingEngineered rat models
Recombinant expressionIn vitro activity of variants
Reporter gene assaysEffect on gene expression
Phenotypic AnalysisIn vitro metabolism studiesKinetic parameters of variants
In vivo pharmacokineticsSystemic exposure differences
Endogenous metabolite profilingBiomarker identification
Clinical TranslationHuman-rat comparative studiesSpecies extrapolation factors
PBPK modelingPrediction of human variability
Population pharmacokineticsQuantification of genetic effects

How can researchers optimize experimental designs to study the role of rat UGT2B17 in drug-drug interactions?

Optimizing experimental designs for drug-drug interaction (DDI) studies involving rat UGT2B17 requires:

  • Substrate and inhibitor selection:

    • Identification of selective substrates for rat UGT2B17

    • Selection of clinically relevant inhibitors and inducers

    • Design of substrate cocktails for simultaneous assessment

  • In vitro experimental approaches:

    • Inhibition kinetics determination (Ki values)

    • Mechanism of inhibition characterization (competitive, non-competitive, uncompetitive)

    • Time-dependent inhibition assessment

    • Induction studies in primary rat hepatocytes

  • Experimental conditions optimization:

    • Pre-incubation steps for time-dependent inhibition

    • Protein concentration and incubation time optimization

    • Selection of appropriate enzyme sources:

      • Recombinant UGT2B17

      • Rat liver microsomes

      • Hepatocytes for comprehensive DDI assessment

  • Data analysis and interpretation:

    • Calculation of inhibition constants (Ki)

    • Determination of IC50 values

    • In vitro-in vivo extrapolation (IVIVE)

    • Physiologically-based pharmacokinetic (PBPK) modeling

  • Translational considerations:

    • Comparison with human UGT2B17 inhibition patterns

    • Species differences in inhibitor potency

    • Scaling factors for preclinical to clinical translation

In one study with human UGTs, R(+)-NAF and S(-)-NAF showed inhibition of UGT1A9 with mean Ki values of 10.0 μM and 11.5 μM, respectively . Similar inhibition studies with rat UGT2B17 would provide valuable comparative data for understanding species-specific drug interactions.

What are the common technical challenges when working with recombinant rat UGT2B17 and how can they be addressed?

Common technical challenges and solutions include:

  • Low expression levels:

    • Optimization of expression vector (codon optimization, strong promoters)

    • Selection of appropriate expression system (insect cells often yield higher UGT expression)

    • Use of chaperones to improve protein folding

    • Optimization of growth conditions and induction parameters

  • Protein instability:

    • Addition of glycerol (10-20%) to storage buffers

    • Use of protease inhibitors during purification

    • Storage at -80°C in small aliquots to avoid freeze-thaw cycles

    • Addition of reducing agents if appropriate

  • Low catalytic activity:

    • Proper activation with alamethicin (5% of microsomal protein concentration)

    • Optimization of buffer conditions and pH

    • Addition of divalent cations (Mg2+, 5-10 mM)

    • UDPGA concentration optimization (typically 4 mM)

  • Analytical detection limitations:

    • Development of sensitive LC-MS/MS methods

    • Use of internal standards for accurate quantification

    • Method optimization for specific glucuronide metabolites

    • Sample preparation techniques to concentrate metabolites

  • Data interpretation challenges:

    • Careful selection of appropriate kinetic models

    • Accounting for non-specific binding to incubation matrices

    • Consideration of potential metabolic pathways beyond glucuronidation

When conducting experiments with recombinant UGTs, preliminary experiments should always be performed to ensure that glucuronides are formed in the linear range of both reaction time and protein concentration , which is essential for accurate kinetic parameter determination.

How should researchers validate the specificity and activity of recombinant rat UGT2B17 preparations?

Comprehensive validation of recombinant rat UGT2B17 preparations should include:

  • Identity confirmation:

    • Western blot analysis with specific antibodies

    • Mass spectrometry peptide fingerprinting

    • N-terminal sequencing

    • RT-PCR confirmation of mRNA expression

  • Purity assessment:

    • SDS-PAGE with Coomassie staining

    • Size exclusion chromatography

    • Determination of specific activity

  • Functional validation:

    • Activity assays with known UGT2B17 substrates

    • Comparison of kinetic parameters with literature values

    • Inhibition profile with selective inhibitors

    • Negative controls (reaction without UDPGA or without enzyme)

  • Cross-reactivity testing:

    • Testing with substrates selective for other UGT isoforms

    • Competition assays with selective substrates

    • Inhibition studies with isoform-selective inhibitors

  • Stability assessment:

    • Activity monitoring over time under storage conditions

    • Thermal stability testing

    • pH stability profile

    • Freeze-thaw stability evaluation

For reliable kinetic characterization, it's essential to determine that the recombinant enzyme preparation retains its activity throughout the experimental procedures, as demonstrated in studies with other UGT isoforms .

What emerging technologies could advance our understanding of rat UGT2B17 structure and function?

Emerging technologies with potential to advance UGT2B17 research include:

  • Advanced structural biology approaches:

    • Cryo-electron microscopy for membrane protein structure determination

    • Hydrogen-deuterium exchange mass spectrometry for conformational dynamics

    • Micro-electron diffraction for crystallography of membrane proteins

    • AlphaFold2 and other AI-based structure prediction tools

  • Systems biology approaches:

    • Multi-omics integration (genomics, proteomics, metabolomics)

    • Network analysis of UGT2B17 interactions

    • Quantitative systems pharmacology modeling

    • Physiologically-based pharmacokinetic modeling

  • Advanced genetic engineering:

    • CRISPR/Cas9 for precise genome editing in rats

    • Base editing for introducing specific mutations

    • Humanized rat models expressing human UGT2B17 variants

    • Conditional knockout models for tissue-specific studies

  • Novel analytical technologies:

    • SWATH-MS for comprehensive UGT quantification

    • High-resolution mass spectrometry for metabolite profiling

    • Imaging mass spectrometry for tissue distribution studies

    • Digital microfluidics for high-throughput enzyme assays

  • Computational approaches:

    • Molecular dynamics simulations of substrate binding

    • Quantum mechanics/molecular mechanics for reaction mechanism studies

    • Machine learning for substrate specificity prediction

    • Virtual screening for selective inhibitors/substrates

These technologies could help address key knowledge gaps, such as the specific contribution of UGT2B17 to rat drug metabolism, its three-dimensional structure, and species differences in substrate specificity that impact translational research.

How might the study of rat UGT2B17 contribute to our understanding of personalized medicine approaches in humans?

Rat UGT2B17 studies can contribute to personalized medicine through:

  • Translational pharmacogenetics:

    • Rat models with varying UGT2B17 genotypes as surrogates for human genetic variation

    • Preclinical assessment of drug response variability

    • Identification of drugs susceptible to UGT2B17-mediated variability in pharmacokinetics

    • Development of predictive biomarkers for drug response

  • Biomarker development:

    • Normalized testosterone glucuronide as a potential urinary biomarker of UGT2B17 activity

    • Correlation of metabolite ratios with genotype

    • Discovery of novel endogenous substrates as activity markers

    • Non-invasive methods to predict UGT2B17 phenotype

  • Drug development implications:

    • Early identification of compounds susceptible to variable metabolism by UGT2B17

    • Prevention of development failures due to unpredictable pharmacokinetics

    • Rational design of drugs less affected by UGT2B17 polymorphisms

    • Development of alternative metabolic pathways for critical medications

  • Clinical application insights:

    • Understanding how UGT2B17 variability affects drug disposition in currently utilized therapeutics like exemestane and vorinostat

    • Development of dosing algorithms based on UGT2B17 genotype/phenotype

    • Identification of potential drug-drug interactions involving UGT2B17

    • Risk stratification for patients based on metabolism profile

Human UGT2B17 variability has led to unexpected pharmacokinetic outcomes resulting in drug development failures, including MK-7246 (showing 25-fold higher systemic exposure in subjects with two gene copies) and PT2385 . Rat studies provide a controlled system to investigate these variations mechanistically.

What interdisciplinary approaches might yield new insights into the evolutionary conservation and divergence of UGT2B17 across species?

Innovative interdisciplinary approaches for comparative UGT2B17 research include:

  • Comparative genomics and phylogenetics:

    • Whole genome analysis across species to track UGT gene evolution

    • Identification of conserved regulatory elements

    • Analysis of selective pressures on UGT2B17 across species

    • Reconstruction of ancestral UGT sequences

  • Evolutionary biochemistry:

    • Functional characterization of UGT2B17 from multiple species

    • Ancestral sequence reconstruction and expression

    • Comparative enzyme kinetics across species

    • Structure-function relationships in relation to evolutionary changes

  • Ecological and environmental toxicology:

    • Species-specific adaptations in detoxification capacity

    • Environmental influences on UGT2B17 evolution

    • Comparative xenobiotic metabolism across ecological niches

    • Dietary influences on UGT substrate specificity evolution

  • Computational biology integration:

    • Molecular dynamics simulations comparing orthologs

    • Machine learning for identifying evolutionary patterns

    • Network analysis of metabolic pathway evolution

    • Structural bioinformatics to map conserved domains

  • Translational implications:

    • Development of species-scaling factors for pharmacokinetic modeling

    • Identification of conserved versus divergent substrate binding sites

    • Selection of appropriate animal models for specific drug classes

    • Prediction of human-specific metabolism based on evolutionary patterns

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.